Open Access

SATB1 protein is associated with the epithelial‑mesenchymal transition process in non‑small cell lung cancers

  • Authors:
    • Natalia Glatzel‑Plucinska
    • Aleksandra Piotrowska
    • Adam Rzechonek
    • Marzenna Podhorska‑Okolow
    • Piotr Dziegiel
  • View Affiliations

  • Published online on: April 28, 2021     https://doi.org/10.3892/or.2021.8069
  • Article Number: 118
  • Copyright: © Glatzel‑Plucinska et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Lung cancer is one of the most frequently diagnosed neoplasms and the leading cause of cancer‑related mortality worldwide. Its predominant subtype is non‑small cell lung cancer (NSCLC), which accounts for over 80% of the cases. Surprisingly, the majority of lung cancer‑related deaths are caused not by a primary tumour itself, but by its metastasis to distant organs. Therefore, it becomes especially important to identify the factors involved in lung cancer metastatic spread. Special AT‑rich binding protein 1 (SATB1) is a nuclear matrix protein that mediates chromatin looping and plays the role of global transcriptional regulator. During the past decade, it has received much attention as a factor promoting tumour invasion. In breast, colorectal and prostate cancers, SATB1 has been shown to influence the epithelial‑mesenchymal transition (EMT) process, which is thought to be crucial for cancer metastasis. The aim of this study was to analyse the possible correlations between the expression of SATB1 and major EMT‑associated proteins in NSCLC clinical samples. Additionally, the impact of EMT induction in NSCLC cell lines on SATB1 mRNA expression was also investigated. Immunohistochemistry was used to assess the expression of SATB1, SNAIL, SLUG, Twist1, E‑cadherin, and N‑cadherin in 242 lung cancer clinical samples. EMT was induced by TGF‑β1 treatment in the A549 and NCI‑H1703 lung cancer cell lines. Changes in gene expression profiles were analyzed using real‑time PCR and Droplet Digital PCR. SATB1 expression was positively correlated with the expression of SNAIL (R=0.129; P=0.045), SLUG (R=0.449; P<0.0001), and Twist1 (R=0.264; P<0.0001). Moreover, SATB1 expression significantly increased after in vitro EMT induction in A549 and NCI‑H1703 cell lines. The results obtained may point to the role of SATB1 as one of the regulators of EMT in NSCLC.

Introduction

With more than 2 million new cases and 1.8 million deaths every year, lung cancer is one of the most frequently diagnosed neoplasms and the leading cause of cancer-related mortality worldwide (1). Unfortunately, despite the development of new forms of treatment, over half of lung cancer patients die within one year of the diagnosis, and the 5-year survival rates do not exceed 18% (2). The predominant lung cancer subtype is non-small cell lung cancer (NSCLC), which accounts for over 80% of cases (3). NSCLCs are highly heterogeneous and have diverse pathological features, thus further histological subtypes are usually distinguished. The most common ones are adenocarcinoma (AC) and squamous cell carcinoma (LSCC) (3).

It is estimated that 90% of NSCLC-related deaths are caused not by the primary tumour itself, but by its metastases to distant organs (4). For lung cancers, the preferable sites for metastasis are the bones, the lungs, the brain, the adrenal glands and the liver, but the exact patterns of metastatic spread are highly dependent on the histological and molecular subtype of the tumour (57). Nevertheless, the presence of distant metastases is an unequivocal hallmark of poor prognosis; for patients with localized NSCLC tumours, the 5-year relative survival rate is approximately 60%, while for disseminated cancers, it decreases 10-fold to only 6% (data from the USA) (8).

The metastasis mechanism, although very complex and not fully understood yet, is commonly thought to rely on two main processes: The epithelial-to-mesenchymal and the mesenchymal-to-epithelial transitions (EMT and MET, respectively). The activation of EMT-promoting traits supports tumour cell invasion and dissemination, while the reverse process, MET, is believed to promote metastatic outgrowth once cancer cells have extravasated and invaded the distant organs (9). During EMT, epithelial cells lose their apical-basal polarity, cell-to-cell junctions and interactions, and contact with the basal membrane (912). They significantly change their morphology and become elongated and spindle-shaped, slowly gaining migration and invasion abilities (912). The expression of key epithelial markers such as E-cadherin or cell junction proteins gradually decreases and the cells start to express proteins typical for the mesenchymal phenotype: N-cadherin, vimentin, and matrix metalloproteinases (MMPs) (911). Physiologically, EMT has been shown to be essential for embryonic development, organogenesis, and tissue repair (12). However, in pathological conditions, as a result of EMT, cancer cells can acquire key metastatic features, including enhanced mobility, invasion ability, and resistance to apoptosis, enabling them to infiltrate the surrounding stroma and to spread through the blood and lymphatic vessels to distant sites (9). In epithelial tumours, EMT is usually chaotic and incomplete, and there are coexisting cells in numerous intermediate states, presenting features of both epithelial and mesenchymal phenotypes (13,14). EMT has long been defined by the loss of the expression of the epithelial marker E-cadherin and the acquisition of the expression of mesenchymal markers vimentin and/or N-cadherin (15). Nowadays, it is known that in cancer cells, EMT is not a binary process, and both epithelial and mesenchymal markers are often co-expressed (15).

The transition from epithelial to mesenchymal phenotype is orchestrated by numerous transcription factors, the most important of which are SNAIL, SLUG, ZEB1, ZEB2, and Twist1 proteins (911). Additionally, it is also regulated epigenetically and at a post-transcriptional level by a variety of different factors, including chromatin modifying enzymes, miRNAs, and long non-coding RNAs (11). Although the relationship between the loss of E-cadherin expression and the increased invasiveness of cancer cells was described for the first time about 30 years ago (16) and the link between EMT and cancer metastasis has been known for almost 20 years (17), the exact molecular pathways associated with EMT in lung cancers and their clinical implications have not yet been fully studied. It has been demonstrated that cigarette smoking, which is considered to be a direct cause of 85–90% of lung cancer cases, can clearly contribute to EMT by decreasing E-cadherin expression (18). It has also been shown that smoking increases the activity of SNAIL and Twist1 in the basal cells of the respiratory epithelium (19). The importance of EMT in NSCLC goes far beyond increasing the metastatic potential and invasiveness of tumour cells. The shift towards a mesenchymal phenotype in these tumours has been linked to the acquired resistance to epidermal growth factor receptor (EGFR) inhibitors, which are the first-line treatment for NSCLC patients harbouring activating EGFR mutations (2022). Interestingly, treatment with EGFR inhibitors was also revealed to be one of the EMT-promoting factors (23). Moreover, as in the case of other cancers, in lung carcinomas, EMT was found to be a strong factor contributing to chemoresistance and radioresistance (2427). Therefore, it becomes especially important to identify and investigate additional factors that might potentially contribute to EMT in NSCLC.

During the past decade, SATB1 (special AT-rich binding protein 1) has received much attention as a factor promoting tumour invasion and metastasis. SATB1 is a nuclear matrix protein that mediates chromatin looping and plays the role of a global transcriptional regulator (28,29). It binds to base-unpairing regions (BURs), the specific AT-rich motifs of double-stranded DNA that may be found every 40,000 DNA base pairs (2830). The binding of SATB1 to BURs provides a nuclear platform necessary for the binding of further transcription factors and chromatin-modifying enzymes, regulates epigenomic modifications and maintains proper nucleosome positioning (30). It binds to distinct genomic regions depending on the cell type. Therefore, it has the ability to regulate whole sets of genes in a tissue-specific manner (28,29,31). SATB1 interactions with transcription activators and repressors are determined by post-transcriptional modifications such as phosphorylation or acetylation (32,33). SATB1 can also be regulated by numerous microRNAs (3440).

Physiologically, SATB1 is expressed in embryonic stem cells and in many adult progenitor cells, for example ameloblasts and osteoblasts (30,41,42). It is essential for embryonic development, and it takes part in processes that require rapid changes in the cell phenotype, such as the differentiation and maturation of thymocytes or skin epithelial cells (28,30). Besides its normal physiological function, SATB1 was also found to be overexpressed in numerous malignancies, including breast, colorectal, prostate, liver, bladder, and ovarian cancers (4349). In these tumours, a high SATB1 level was clearly associated with an aggressive phenotype, the presence of metastasis and a poor patient prognosis (4349).

In most of the abovementioned cancers, a high SATB1 level was also shown to influence the EMT process and to impact the expression of EMT-related proteins. In breast cancer cells, SATB1 was found to promote the mesenchymal phenotype by upregulating vimentin and N-cadherin, as well as downregulating the key epidermal markers claudin-1, β-catenin, and E-cadherin (43). It also stimulated the expression of the most crucial EMT-associated transcription factors: SNAIL and Twist1 (50). It was shown to play a role in the induction of chemotherapy-related EMT (36), and to increase the number of breast cancer stem cells (BCSCs) within the tumours (50). Moreover, SATB1 depletion in MDA-MB-231, a highly aggressive and tumorigenic breast cancer cell line, was demonstrated to reverse the EMT process and to significantly change the phenotype of the cells, restoring their polarization and acinar-like morphology (43). In colorectal tumours, SATB1 expression was found to be positively correlated with the expression of vimentin, and negatively with the expression of the epithelial proteins E-cadherin and CK20 (51). SATB1 knockdown in colorectal cancer cell lines was shown to affect the expression of EMT-related proteins, including E-cadherin, N-cadherin, SLUG, and Twist1 (52). Links between SATB1 expression and the EMT process were also observed in prostate cancer. The studies on the loss-of-function models revealed that SATB1 expression was required to maintain the invasive phenotype of prostate cancer cells, and that its knockdown significantly inhibited cell growth, proliferation, and invasion rates (45,5355). Moreover, its silencing was shown to increase E-cadherin expression, and to restore the anchorage-dependent growth of the cells together with their polarized morphology (45,55). SATB1 associations with the EMT process have been revealed in liver cancer as well, where SATB1 was shown to influence the expression of more than 100 genes related to tumour progression and metastasis, including genes coding for EMT-related proteins such as SNAIL, SLUG, Twist1, vimentin, and E-cadherin (46). Finally, the downregulation of E-cadherin and the upregulation of SNAIL, SLUG, and vimentin as a result of SATB1 overexpression were also observed in bladder cancer cells (56).

Although numerous studies have revealed that SATB1 may have a significant impact on cancer progression, metastasis, and the EMT process, its role in NSCLC remains ambiguous and not fully understood. It is known that SATB1 is necessary for proper lung development during embryogenesis, and that its depletion in mice is lethal (57). It has also been observed that the SATB1 level is elevated in human respiratory epithelial cells (58,59), whereas in the lung alveoli its expression is rather low (5961). To date, the most comprehensive study concerning the role of SABT1 in NSCLC was published in 2011 by Selinger et al, who were the first to discover that SATB1 expression is associated with lung squamous cell carcinoma (LSCC) tumour histology and a poor degree of histological differentiation in the whole NSCLC study cohort (58). Surprisingly, they also observed that the loss of SATB1 expression was a negative prognostic factor for LSCC patients (58). In 2016, Huang et al assessed SATB1 expression in adenocarcinoma (AC) samples only, and they confirmed that an elevated SATB1 level was related to a higher tumour grade (60). In our recent study, we decided to analyse SATB1 expression in particular NSCLC subtypes separately (59). We found the SATB1 level to be significantly higher in LSCCs in comparison to AC specimens (59), confirming previous findings made by Selinger et al (58). Moreover, while in ACs the expression of SATB1 was associated with a poor degree of histological differentiation, in LSCCs the level of SATB1 was increased in well differentiated tumours (59). We also observed the negative impact of a decreased SATB1 expression on NSCLC patient survival (59). However, the results were statistically significant only for the whole study cohort, and no association was noted between SATB1 expression and the survival of AC patients (59). For LSCC, the results were on the verge of statistical significance (59). These results suggest that in NSCLC, the function of SATB1 may be highly dependent on the exact tumour histology.

There are no reports available on the possible impact of SATB1 expression on the EMT process in NSCLC. Therefore, the aim of this study was to analyse the possible correlations between the expression of SATB1 and major EMT-associated proteins (E-cadherin, N-cadherin, SNAIL, SLUG, and Twist1) in NSCLC clinical samples. Additionally, we also investigated the impact of transforming growth factor (TGF-β1) exposure, which is commonly used to induce EMT in vitro, on SATB1 mRNA expression in AC and LSCC cell lines.

Materials and methods

Patient cohort

The present study was approved by the Bioethics Commission at the Wroclaw Medical University in Poland (approval no. KB-632/2017). A total of 262 NSCLC and adjacent non-malignant lung tissue (NMLT) samples were collected from patients treated at the Lower Silesian Centre of Lung Diseases in Wroclaw during the years 2007–2016. The study group consisted of 150 adenocarcinomas (AC), 92 squamous cell carcinomas (LSCC), and 20 non-malignant lung tissue (NMLT) samples. For each sample, formalin-fixed paraffin-embedded (FFPE) tissue blocks were prepared. The histological type of the tumours was assessed using the World Health Organization Classification (3) by two independent pathologists, and was additionally confirmed by immunohistochemical staining for the marker proteins TTF-1 (AC marker) and p63 (LSCC marker). The pTNM classification was made according to the recommendations of the International Association for the Study of Lung Cancer (IASLC) (62). The clinical, pathological, and survival data were obtained from the hospital archives and are listed in Table I.

Table I.

Clinicopathological data of the NSCLC patients.

Table I.

Clinicopathological data of the NSCLC patients.

ParametersAll cases (N=242) n (%)AC (N=150) n (%)LSCC (N=92) n (%)
Sex
  Male146 (60.3)85 (56.7)61 (66.3)
  Female  96 (39.7)65 (43.3)31 (33.7)
Age
  Mean66.22±7.6565.87±8.1466.81±6.77
  Range44-8444-8444-82
Malignancy grade
  G1  3 (1.2)  3 (2.0)  0 (0.0)
  G2152 (62.8)73 (48.7)79 (85.9)
  G3  87 (36.0)74 (49.3)13 (14.1)
Tumour size
  pT1  76 (31.4)56 (37.3)20 (21.7)
  pT2124 (51.2)66 (44.0)58 (63.0)
  pT3  23 (9.5)11 (7.3)12 (13.0)
  pT4  5 (2.1)  4 (2.7)  1 (1.1)
  No data  14 (5.8)13 (8.7)  1 (1.1)
Lymph nodes
  pN0147 (60.7)83 (55.3)64 (69.6)
  pN140 (16.5)23 (15.3)17 (18.5)
  pN241 (16.9)31 (20.7)10 (10.9)
  No data14 (5.8)13 (8.7)  1 (1.1)
Stage
  I104 (43.0)64 (42.7)40 (43.5)
  II  77 (31.8)37 (24.7)40 (43.5)
  III  45 (18.6)34 (22.7)11 (12.0)
  IV  2 (0.83)  2 (1.3)  0 (0.0)
  No data  14 (5.8)13 (8.7)  1 (1.1)
Overall survival
  Deceased  95 (39.3)63 (42.0)32 (34.8)
  Alive146 (60.3)86 (57.3)60 (65.2)
  No data  1 (0.41)  1 (0.67)  0 (0.0)

[i] NSCLC, non-small cell lung cancer; AC, adenocarcinoma; LSCC, squamous cell carcinoma. Age is expressed in years, all other data are expressed as n (%).

Cell lines

The NCI-H1703 (LSCC) and A549 (AC) cell lines were obtained from the American Type Culture Collection (ATCC). The cell culture media used were RPMI-1640 (for the NCI-H1703 cell line) and F12K (for the A549 cell line). All of the media were additionally supplemented with L-glutamine up to a final concentration of 2 mM, and with fetal bovine serum, up to a final concentration of 10%. All of the cell culture media and reagents were purchased from Sigma-Aldrich; Merck KGaA. Cell culture conditions were as follows: Temperature 37°C and 5% CO2 concentration.

Tissue microarrays (TMAs)

Tissue microarrays (TMAs) were created using the TMA Grand Master (3DHistech) automatic tissue microarrayer. From each FFPE tissue block, standard 4-µm-thick paraffin sections were cut and hematoxylin and eosin (H&E) stained. Then, the prepared slides were scanned with a Pannoramic MIDI II (3DHistech) histological scanner. Representative spots for the TMAs (three spots with 1.5 mm diameter from each FFPE block) were selected by a qualified pathologist from the digital slides with the use of the Case Viewer (3DHistech) software. Finally, TMAs were created using the TMA Grand Master system, according to the manufacturer's instructions.

Immunohistochemistry (IHC)

Immunohistochemical reactions were performed on 4-µm-thick paraffin sections using DAKO Autostainer Link48 (Dako; Agilent Technologies, Inc.). First, deparaffinization, rehydration, and antigen retrieval were performed using EnVision FLEX Target Retrieval Solution (pH 9.0, 20 min, 97°C) in PTLink (Dako; Agilent Technologies, Inc.). In order to block the activity of endogenous peroxidase, the sections were incubated in EnVision FLEX Peroxidase-Blocking Reagent (5 min at room temperature). Afterwards, primary antibodies directed against E-cadherin (RTU; cat. no. IR059; Dako; Agilent Technologies, Inc.), N-cadherin (1:50; cat. no. M3613; Dako; Agilent Technologies, Inc.), SNAIL (1:400; cat. no. 13099-1-AP; ProteinTech), SLUG (1:50, cat. no. 166476; SantaCruz Biotechnology, Inc.), and Twist1 (1:500; cat. no. ab50581; Abcam) were applied for 20 min at room temperature. Then, the slides were incubated with EnVision FLEX/HRP (20 min). Next, 3,3′-diaminobenzidine (DAB, Dako; Agilent Technologies, Inc.) was utilized as the peroxidase substrate, and the sections were incubated for 10 min at room temperature. Additionally, all the sections were counterstained with FLEX Hematoxylin for 5 min at room temperature. After dehydration in graded ethanol concentrations (70%, 96%, absolute) and in xylene, the slides were mounted in Dako Mounting Medium using Coverslipper (Dako; Agilent Technologies, Inc.). The primary antibodies were diluted in FLEX Antibody Diluent.

Evaluation of IHC reactions

IHC slides were scanned with the Pannoramic MIDI II (3DHistech) histological scanner and evaluated using the QuantCenter (3DHistech) digital image analysis software. The cells of interest (cancer cells or normal lung alveolar cells) were distinguished from the other cellular components using the PatternQuant (3DHistech) software module. Then, the CellQuant (3DHistech) module was used to determine the percentage of positive cells and the intensity of the reaction among the selected tissue compartments only. The expression levels of SATB1 and the Ki-67 proliferative index were evaluated as previously described (59). The membranous expression of E-cadherin and N-cadherin was assessed using a scale ranging from 0 to 3, based on the percentage of positive cells and the intensity of the staining (Table II).

Table II.

Scoring system used for the evaluation of membranous staining.

Table II.

Scoring system used for the evaluation of membranous staining.

ScorePercentage of positive cells and intensity of staining
0No staining is observed or staining is observed in <10% of the tumour cells
1A faint membrane staining is observed in >10% of the tumour cells
2A weak or moderate, complete membrane staining is observed in >10% of the tumour cells
3A strong, complete membrane staining is observed in >10% of the tumour cells

SNAIL, SLUG, and Twist1 protein expression levels were assessed using the Allred scale. This scoring system is calculated by adding a number representing the proportion of positive cells (0–5) to a number reflecting the intensity of the staining (0–3) (63). The final score value ranges from 0 to 8, where 0 indicates no positive cells, and 8 indicates more than 66% of highly positive cells (Table III). The Allred scale can be used to evaluate both nuclear and cytoplasmic stainings.

Table III.

Allred scoring system (63).

Table III.

Allred scoring system (63).

APercentage of positive cellsBIntensity of staining
0No positive cells0No detectable staining
1<1%1Weak staining
21-10%2Moderate staining
310-33%3Strong staining
433-66%
5>66%

[i] The final score was calculated as a sum of the factors A and B. It fits within a range from 0 to 8.

In vitro EMT induction

To induce EMT in cell culture conditions, A549 (1×105/well) and NCI-H1703 (2×105/well) cells were seeded in 6-well plates and maintained for 24 h in complete culture medium (F12K and RPMI-1640, respectively), supplemented with 5% FBS. Further, the medium was replaced with a fresh one, and the cells were treated with 5 ng/ml TGF-β1 (Merck KGaA) and cultured for the next 72 h. Every 24 h, cell lysates were prepared and subjected to further analysis. Additionally, the cells were examined under a light microscope to track the morphological changes.

RNA isolation and cDNA synthesis

Total RNA from the NCI-H1703 and A549 cell lines was isolated using the GeneMATRIX® Universal RNA/miRNA Purification Kit (EURx, Gdansk, Poland), according to the manufacturer's handbook. For each sample, 500 ng of total RNA was transcribed into cDNA using the iScript™ Reverse Transcription Supermix for RT-qPCR (Bio-Rad, Laboratories, Inc.) and the C1000 Touch Thermal Cycler (Bio-Rad, Laboratories, Inc.). The reaction conditions were as follows: Priming for 5 min at 25°C, reverse transcription for 20 min at 46°C, and final inactivation of reverse transcriptase for 1 min at 95°C.

Real-time PCR

The real-time PCR method was used to determine the relative levels of CDH1, CDH2, SNAI1, SNAI2, and Twist1 mRNA expression in the NCI-H1703 and A549 cell lines before and after EMT induction. The reactions were performed using the 7900 Real Time PCR System (Applied Biosystems; Thermo Fisher Scientific, Inc.) and the iTaq™ Universal Probes Supermix (Bio-Rad, Laboratories, Inc.), according to the manufacturer's instructions. The TaqMan probes used are listed in Table IV. SDHA an endogenous control gene, was further used for normalisation purposes. The reactions were carried out in triplicates under the following conditions: Initial denaturation at 94°C for 2 min, followed by 45 cycles of denaturation (94°C, 15 sec), and annealing with elongation (60°C, 1 min). The relative mRNA expression levels were calculated using the ΔΔCt method.

Table IV.

TaqMan probes used in the experiment.

Table IV.

TaqMan probes used in the experiment.

Protein nameGene symbolTaqMan probe
E-cadherinCDH1Hs01023894_m1
N-cadherinCDH2Hs00983056_m1
SNAILSNAI1Hs00195591_m1
SLUGSNAI2Hs00950344_m1
Twist1Twist1Hs01675818_s1
SDHASDHAHs99999903_m1
Droplet digital PCR

The droplet digital PCR method was used to determine the absolute number of SATB1 mRNA copies in the analysed cell lines before and after EMT induction. The reaction mixture contained 3.33 µl of the RT product, 1 µl of the SATB1-specific TaqMan probe Hs00962580_m1 (Thermo Fisher Scientific, Inc.), 5.67 µl of the molecular biology-grade water, and 10 µl of the ddPCR™ Supermix for Probes (Bio-Rad Laboratories, Inc.). A total of 20 µl of the reaction mixture was loaded onto a plastic cartridge with 70 µl of the Droplet Generation Oil for Probes (Bio-Rad Laboratories, Inc.) and inserted into the QX200 Droplet Generator (Bio-Rad Laboratories, Inc.). The droplets obtained from each sample were then transferred to a 96-well PCR plate (Eppendorf). PCR amplifications were carried out in the C1000 Touch Thermal Cycler (Bio-Rad Laboratories, Inc.) under the following conditions: Enzyme activation at 95°C for 10 min, followed by 40 cycles of denaturation (94°C, 30 sec), annealing/extension (60°C, 1 min), and a final enzyme deactivation at 98°C for 10 min. Finally, the plate was loaded onto the QX200 Droplet Reader (Bio-Rad Laboratories, Inc.) and read automatically. The quantification of the SATB1 mRNA is presented as the total number of copies in the reaction mixture.

Statistical analysis of the results

All the experiments, except for the IHC stainings, were performed at least in triplicates. The results were analysed with the use of Prism 8.0 software (GraphPad Software) and Statistica 13 (StatSoft, Krakow, Poland) statistical software. The Shapiro-Wilk test was utilized to determine if the sample data were normally distributed. To compare the groups of data, a non-parametric Mann-Whitney U test or a parametric Student's test was used. The correlations between the analysed parameters were checked using the Spearman's rank correlation test. The survival times were determined by using the Kaplan-Meier method, and the significance of differences was determined by a log-rank test. All the results were considered statistically significant when P<0.05.

Results

E-cadherin is significantly overexpressed in NSCLC compared to NMLT samples

Membranous E-cadherin expression was observed both in cancer cells (Fig. 1B) and non-malignant lung tissues-respiratory epithelial cells (Fig. 1A) and pneumocytes (Fig. 1A). Strong or moderate (score ≥2) E-cadherin staining was present in 99.17% of the analysed NSCLC cases, and the mean score values were significantly higher in NSCLC compared to the NMLT samples (2.96±0.24 vs. 1.10±0.64; P<0.0001). No significant correlations between the expression of E-cadherin and the patient clinicopathological data were noted (Table SI).

N-cadherin level is related to the stage of the disease, and is correlated negatively with tumour grade

N-cadherin expression (score ≥1) was observed in the membranes of cancer cells in 7.85% of the analysed NSCLC cases (Fig. 1D). In NMLT samples, no N-cadherin staining was detected (Fig. 1C). In the whole study cohort, mean N-cadherin scores were significantly higher in G1-G2 tumours compared to G3 ones (0.19±0.58 vs. 0.03±0.24; P=0.02; Table SII). A similar relationship was also observed in AC cases analysed separately (0.17±0.60 vs. 0.01±0.12; P=0.05; Table SII). Moreover, N-cadherin overexpression was associated with a more advanced stage of the disease, both in the whole study cohort (0.17±0.51 in stage II–IV tumours vs. 0.05±0.35 in stage I tumours; P=0.007; Table SII), and in the LSCC subtype (0.31±0.68 in stage II–IV tumours vs. 0.05±0.32 in stage I tumours; P=0.02; Table SII).

In LSCC, SNAIL overexpression was associated with an advanced stage of the disease and the presence of lymph node metastasis

SNAIL was highly expressed in the cytoplasm of cancer cells (Fig. 1F) and, less abundantly, in the cytoplasm of normal lung macrophages (Fig. 1E). A total of 82.64% of NSCLC specimens were SNAIL-positive (Allred score >3). No SNAIL expression was observed in non-malignant lung alveoli (Fig. 1E) and bronchial epithelium. Although mean SNAIL scores did not differ between particular NSCLC subtypes (Table V), the exact expression patterns and associations with the clinicopathological data seemed to be dependent on the tumour histology. While in AC there was no relationship between SNAIL expression and the presence of lymph node metastasis, SNAIL was significantly overexpressed in pN1 and pN2 tumours compared to pN0 ones (5.22±1.05 vs. 4.69±1.00; P=0.02; Table V) in LSCC. Additionally, in LSCC, an elevated SNAIL level was associated with a more advanced stage of the disease; in stage II–IV tumours mean SNAIL scores were significantly higher compared to stage I (5.04±1.04 vs. 4.63±1.01; P=0.04; Table V). Similar associations between SNAIL expression and the stage of the disease were also observed in the whole study cohort, but not in the AC subtype analysed separately (Table V).

Table V.

SNAIL expression and clinicopathological data of NSCLC patients.

Table V.

SNAIL expression and clinicopathological data of NSCLC patients.

All cases (N=242)AC (N=150)LSCC (N=92)



ParametersScore ± SDP-valueScore ± SDP-valueScore ± SDP-value
Histological type
  AC4.693±1.3260.37
  LSCC4.848±1.037
Age (years)
  ≤654.596±1.2560.084.536±1.3990.194.700±0.96610.25
  >654.880±1.187 4.827±1.253 4.962±1.084
Sex
  Male4.733±1.2280.894.706±1.3520.864.770±1.0390.49
  Female4.758±1.209 4.677±1.300 4.933±0.9803
Malignancy grade
  G1, G24.794±1.2150.384.684±1.3780.994.899±1.0330.16
  G34.678±1.244 4.703±1.279 4.538±1.050
Tumour size
  pT14.605±1.2760.254.500±1.3880.274.900±0.85220.95
  pT2-pT44.803±1.218 4.765±1.325 4.845±1.091
Lymph nodes
  pN04.671±1.2390.154.656±1.3830.714.692±0.99880.02
  pN1, pN24.914±1.185 4.759±1.228 5.222±1.050
Stage
  I4.529±1.2380.024.469±1.3680.164.625±1.0050.04
  II–IV4.911±1.216 4.822±1.326 5.039±1.038

[i] NSCLC, non-small cell lung carcinoma; AC, adenocarcinoma; LSCC, squamous cell carcinoma. Significant P-values are provided in bold.

Relationships between SLUG expression and the patient clinicopathological data are dependent on protein localisation (nuclear vs. cytoplasmic) and tumour histology

SLUG staining was detected in the cytoplasm and nuclei of cancer cells (Fig. 1H) and in the cytoplasm of normal lung macrophages (Fig. 1G). We observed cytoplasmic SLUG (SLUG C) expression (Allred score >3) in 75.62% of NSCLC cases, whereas nuclear expression (SLUG N; Allred score >3) was present in 51.24% of the samples. In 50.83% of the analysed cases, SLUG was expressed both in the cytoplasm and nuclei of the cells.

SLUG C expression patterns differed visibly depending on the tumour histology. In AC, mean SLUG C scores were significantly higher in G3 tumours compared to G1 and G2 ones (5.70±0.95 vs. 5.32±1.17; P=0.05; Table VI). The association between SLUG C expression and the tumour malignancy grade was also present in the whole study cohort (Table VI). At the same time, in LSCC, the level of SLUG C seemed to be positively associated with the tumour size (5.44±1.02 in pT2-pT4 tumours vs. 4.75±1.58 in pT1 tumours; P=0.04; Table VI) and the stage of the disease (5.57±0.88 in stage II–IV tumours vs. 4.92±1.44 in stage I tumours; P=0.02; Table VI).

Table VI.

Cytoplasmic SLUG expression and clinicopathological data of the NSCLC patients.

Table VI.

Cytoplasmic SLUG expression and clinicopathological data of the NSCLC patients.

All cases (N=242)AC (N=150)LSCC (N=92)



ParametersScore ± SDP-valueScore ± SDP-valueScore ± SDP-value
Histological type
  AC5.507±1.0790.11
  LSCC5.293±1.191
Age (years)
  ≤655.266±1.3030.165.333±1.2330.215.150±1.4240.47
  >655.556±0.9408 5.654±0.9105 5.404±0.9754
Sex
  Male5.370±1.1510.535.506±1.0540.785.180±1.2580.29
  Female5.505±1.090 5.508±1.120 5.500±1.042
Malignancy grade
  G1, G25.303±1.1810.035.316±1.1690.055.291±1.2000.94
  G35.644±0.9880 5.703±0.9469 5.308±1.182
Tumour size
  pT15.237±1.2840.095.411±1.1250.314.750±1.5850.04
  pT2-pT45.487±1.048 5.531±1.073 5.437±1.024
Lymph nodes
  pN05.453±1.1450.495.615±1.0190.095.215±1.2810.37
  pN1, pN25.370±1.089 5.315±1.163 5.481±0.9352
Stage
  I5.375±1.2320.885.656±0.99550.084.925±1.4390.02
  II–IV5.427±1.053 5.329±1.155 5.569±0.8776

[i] NSCLC, non-small cell lung carcinoma; AC, adenocarcinoma; LSCC, squamous cell carcinoma. Significant P-values are provided in bold.

SLUG N was significantly overexpressed in LSCC compared to AC tumours (4.17±1.04 vs. 3.56±0.75; P<0.001; Table VII). In LSCC, the level of SLUG N was positively associated with the tumour size (4.27±1.04 in pT2-pT4 tumours vs. 3.80±1.01 in pT1 tumours; P=0.04; Table VII) and the stage of the disease (4.33±0.97 in stage II–IV tumours vs. 3.95±1.11 in stage I tumours; P=0.02; Table VII). A positive relationship between tumour size and SLUG N expression was also observed in the whole study cohort, but not in the AC subtype (Table VII). In AC, SLUG N expression level was revealed to be associated with the patient sex: It was significantly higher in men than in women (3.69±0.76 vs. 3.39±0.70, respectively; P=0.01; Table VII).

Table VII.

Nuclear SLUG expression and clinicopathological data of the NSCLC patients.

Table VII.

Nuclear SLUG expression and clinicopathological data of the NSCLC patients.

All cases (N=242)AC (N=150)LSCC (N=92)



ParametersScore ± SDP-valueScore ± SDP-valueScore ± SDP-value
Histological type
  AC3.560±0.7462 <0.001
  LSCC4.174±1.044
Age (years)
  ≤653.725±0.96100.213.449±0.75800.094.200±1.0910.47
  >653.850±0.8833 3.654±0.7273 4.154±1.017
Sex
  Male3.884±0.94330.053.694±0.75630.014.148±1.1080.29
  Female3.642±0.8619 3.385±0.7003 4.200±0.9248
Malignancy grade
  G1, G23.858±0.97660.223.474±0.70190.184.228±1.0620.94
  G33.678±0.7996 3.649±0.7840 3.846±0.8987
Tumour size
  pT13.526±0.79120.0023.429±0.68380.123.800±1.0050.04
  pT2-pT43.908±0.9653 3.593±0.7710 4.268±1.041
Lymph nodes
  pN03.752±0.86620.353.542±0.67930.674.062±1.0140.37
  pN1, pN23.877±1.017 3.593±0.8582 4.444±1.086
Stage
  I3.673±0.85270.093.500±0.59090.853.950±1.1080.02
  II–IV3.871±0.9792 3.548±0.8505 4.333±0.9730

[i] NSCLC, non-small cell lung carcinoma; AC, adenocarcinoma; LSCC, squamous cell carcinoma. Significant P-values are provided in bold.

Twist1 is expressed mainly in LSCC tumours, and its expression levels are correlated with the stage of the disease

Twist1 expression was observed in the nuclei of cancer cells (Fig. 2C) and in the nuclei of infiltrating lymphocytes (Fig. 2B). In NMLT samples, no Twist1 staining was detected (Fig. 2A). A total of 12.81% of the analysed NSCLC samples were Twist1-positive (Allred score >3). Twist1 expression levels were related to the tumour histology; in LSCC, mean score values were significantly higher compared to AC (3.50±1.19 vs. 2.74±0.62, respectively; P<0.001; Table SIII). Moreover, in LSCC, Twist1 expression was positively associated with the stage of the disease (3.73±1.33 in stage II–IV tumours vs. 3.20±0.94 in stage I tumours; P=0.04; Table SIII). Inversely, in AC, a negative relationship between Twist1 level and the tumour size was observed; in pT2-pT4 tumours, Twist1 expression was significantly decreased compared to pT1 ones (2.62±0.68 vs. 2.86±0.55, respectively; P=0.02; Table SIII).

SATB1 scores are positively correlated with SNAIL, SLUG, and Twist1 expression

Further analysis revealed that SNAIL, SLUG, and Twist1 expression levels were positively correlated with SATB1 scores, as assessed in our previous study (59). In the whole study cohort, we observed a moderate positive correlation between the expression of SATB1 and SLUG N (R=0.449; P<0.0001; Table VIII; Fig. 3G), and a low positive correlation between the expression of SATB1 and SLUG C (R=0.288; P<0.0001; Table VIII; Fig. 3D). SATB1 scores were also correlated positively with Twist1 and SNAIL levels (R=0.264; P<0.0001 and R=0.129; P<0.045, respectively; Table VIII; Fig. 3J and A, respectively).

Table VIII.

Correlations between the expression of SATB1 and E-cadherin, N-cadherin, SNAIL, SLUG, and Twist1 proteins.

Table VIII.

Correlations between the expression of SATB1 and E-cadherin, N-cadherin, SNAIL, SLUG, and Twist1 proteins.

NSCLCACLSCC



ProteinSpearman's RP-valueSpearman's RP-valueSpearman's RP-value
E-cadherin0.1090.0900.0690.4000.1430.174
N-cadherin0.0800.215−0.0040.9630.1260.232
SNAIL0.1290.0450.0920.2610.1520.148
SLUG C0.288 <0.00010.294 <0.0010.403 <0.0001
SLUG N0.449 <0.00010.365 <0.00010.424 <0.0001
Twist10.264 <0.00010.2180.0080.1440.169

[i] SATB1, special AT-rich binding protein 1; NSCLC, non-small cell lung carcinoma; AC, adenocarcinoma; LSCC, squamous cell carcinoma. C, cytoplasmic; N, nuclear. Significant P-values are provided in bold.

Positive correlations between the expression levels of SATB1 and SLUG N on the one hand, and the expression levels of SATB1 and SLUG C on the other were also observed in the AC and LSCC subtypes analysed separately. R values were especially high in LSCC: 0.424 for SATB1/SLUG N and 0.403 for SATB1/SLUG C correlation (Table VIII; Fig. 3). However, a significant correlation between SATB1 and Twist1 expressions was found only in AC (R=0.218; P=0.008; Table VIII; Fig. 3K).

Expression of EMT markers is positively correlated with Ki67 scores

The expression of all of the markers analysed in the whole study cohort was found to be positively correlated with Ki67 scores, as assessed in our previous study (59). The correlation coefficients were particularly high for SNAIL and SLUG N (R=0.372; P<0.0001 and R=0.347; P<0.0001, respectively; Table IX). Significant positive correlations between the expression of Ki67 and E-cadherin, N-cadherin, SNAIL, SLUG C, and SLUG N were also detected in the AC subtype (Table IX). In LSCC, Ki67 expression correlated moderately with the expression of SNAIL and SLUG C (R=0.449; P<0.0001 and R=0.410; P<0.0001, respectively; Table IX), and more weakly with the expression of SLUG N and Twist1 (R=0.284; P<0.006 and R=0.299; P<0.004, respectively; Table IX). Interestingly, in LSCC, no associations between Ki67, E-cadherin, and N-cadherin levels were observed (Table IX).

Table IX.

Correlations between the expression of E-cadherin, N-cadherin, SNAIL, SLUG, and Twist1 and the Ki-67 proliferative index.

Table IX.

Correlations between the expression of E-cadherin, N-cadherin, SNAIL, SLUG, and Twist1 and the Ki-67 proliferative index.

NSCLCACLSCC



ProteinSpearman's RP-valueSpearman's RP-valueSpearman's RP-value
E-cadherin0.2020.0020.2490.0020.0600.569
N-cadherin0.1960.0020.1730.0350.1820.082
SNAIL0.372 <0.00010.351 <0.00010.449 <0.0001
SLUG C0.228 <0.0010.1980.0150.410 <0.0001
SLUG N0.347 <0.00010.2350.0040.2840.006
Twist10.2150.001−0.0360.6620.2990.004

[i] NSCLC, non-small cell lung carcinoma; AC, adenocarcinoma; LSCC, squamous cell carcinoma. C, cytoplasmic; N, nuclear. Significant P-values are provided in bold.

N-cadherin expression is a negative prognostic factor for LSCC patients

In order to determine the impact of N-cadherin expression on patient survival, Kaplan-Meier's survival curves were compared using the log-rank (Mantel-Cox) test. The results revealed that an elevated N-cadherin level (score ≥1) was a significant negative prognostic factor for LSCC patients (P=0.0093; Fig. 4C), but not for AC patients (P=0.7235; Fig. 4B). N-cadherin expression lacked a prognostic significance in the whole study cohort as well (P=0.8746; Fig. 4A).

Apart from N-cadherin, none of the markers analysed (E-cadherin, SNAIL, SLUG, or Twist1) was observed to have a significant impact on NSCLC patient survival.

TGF-β1 treatment successfully induces EMT in NSCLC cell line

To determine whether SATB1 expression changes during the EMT process, we induced EMT in vitro in the A549 (AC) and NCI-H1703 (LSCC) cell lines using TGF-β1. Forty eight hours after exposure to TGF-β1, we observed significant morphological changes in the cultured cells, which indicated a shift from an epithelial to a mesenchymal phenotype. A549 cells lost their usual cobblestone-like morphology (Fig. 5A) and became elongated and spindle-shaped. Moreover, they grew dispersed and showed reduced cell-to-cell contact (Fig. 5B). In NCI-H1703 cells, due to their normally elongated shape (Fig. 5C), morphological changes were less evident but still noticeable. Additionally, after EMT induction, these cells developed numerous cytoplasmic extensions, probably associated with an enhanced cell motility (64) (Fig. 5D).

These morphological transformations were accompanied with a gradual shift from epithelial to mesenchymal gene expression profiles. To track these changes, we performed real-time PCR assays and analysed the expression of key EMT-related genes, CDH1, CDH2, SNAI1, SNAI2, and TWIST1 (coding for E-cadherin, N-cadherin, SNAIL, SLUG, and Twist1, respectively). After EMT induction, in the A549 cell line we observed about a 10-fold decrease in CDH1 expression (Fig. 6A) and a significant increase in the expression of CDH2, SNAI1 and SNAI2 (Fig. 6B-D). In the NCI-H1703 cells, the expression levels of CDH1 remained unchanged after TGF-β1 exposure (Fig. 7A), while the expression of CDH2, SNAI1, and SNAI2 increased significantly (Fig. 7B-D). In both cell lines, EMT induction had no effect on Twist1 mRNA levels (Figs. 6E and 7E).

SATB1 expression in A549 and NCI-H1703 cell lines is significantly increased after EMT induction

In order to investigate whether SATB1 expression in NSCLC cell lines changes during EMT, we used the Droplet Digital PCR method to assess SATB1 mRNA levels in the cell culture lysates obtained from the cells after TGF-β1 treatment and from the control ones. In both of the analysed cell lines (A549 and NCI-H1703), induction of EMT resulted in a significant increase in SATB1 expression levels. However, the exact SATB1 expression patterns were different depending on the cell line.

In the control (untreated A549 cells), SATB1 mRNA level gradually increased during the time of the culture, reaching its maximum value (62.00±40.51 mRNA copies) after 72 h. Twenty hours after TGF-β1 exposre, SATB1 expression in EMT cells significantly increased compared to the control ones (67.11±28.60 vs. 15.36±11.79; P=0.0001; Fig. 8A). Forty-eight hours after the EMT induction, SATB1 mRNA level was still slightly elevated, but not significantly different from the control (59.11±48.51 vs. 36.67±25.22; ns; Fig. 8A). After 72 h, SATB1 expression in EMT cells decreased to a control level (72.67±32.95 mRNA copies/sample).

Untreated NCI-H1703 cells showed a rather low SATB1 mRNA level (no more than 30 mRNA copies/sample), which remained stable during the 72 h of culture. Yet, 24 h after the EMT induction, we observed a sharp increase in SATB1 expression that lasted for about 48 h after TGF-β1 exposre; 24 and 48 h after TGF-β1 administration, SATB1 mRNA levels were significantly elevated compared to the control (103.70±57.29 vs. 27.13±14.94 after 24 h; P=0.0029; 105.40±73.85 vs. 26.13±9.48 after 48 h; P=0.0098; Fig. 8B). Yet, 72 h after EMT induction, SATB1 expression dropped to a level comparable to the control cells (16.29±10.80 vs. 13.58±7.32; ns; Fig. 8B).

Discussion

The epidermal-mesenchymal transition (EMT) process is commonly thought to be one of the main mechanisms underlying cancer metastasis by enabling cancer cells to acquire an invasive phenotype (9,15,17). Although the complex network of molecular factors orchestrating EMT has been extensively studied over the past 20 years, there are still unanswered questions and challenging topics to be explored. In recent years, there has been a growing interest in special AT-rich binding protein 1 (SATB1), a potent transcriptional regulator whose role in EMT was confirmed, among others, in breast, colorectal, prostate and bladder cancers (65). In these neoplasms, SATB1 was demonstrated not only to be associated with a high tumour malignancy and a poor patient prognosis, but also to promote an invasive, mesenchymal phenotype of the cells (65). In cancer cell lines, it was found to stimulate the expression of key mesenchymal markers such as vimentin and N-cadherin, as well as the most important EMT-associated transcription factors: SNAIL, SLUG, and Twist1 (43,46,50,52,56). It was also demonstrated to act as a repressor of the expression of epithelial proteins, including claudin-1 and E-cadherin (43,46,56). SATB1 depletion in aggressive, tumorigenic cancer cell lines was sufficient not only to decrease cell proliferation rates, invasiveness, and resistance to apoptosis, but also to restore their polarization and anchorage-dependent growth (43,45,5255,6668). These reports suggest that, in specific conditions, SATB1 may function as a specific ‘trigger’ of the malignant phenotype, clearly contributing to EMT and cancer metastasis.

Unfortunately, little is known about the role of SATB1 in the progression of non-small cell lung cancer (NSCLC), which is one of the most common and malignant human cancers. Previous reports are ambiguous and indicate that the function of SATB1 in NSCLC is highly dependent on the tumour histology [adenocarcinoma (AC) vs. squamous cell carcinoma (LSCC)] (5860). To date, the possible role of SATB1 in EMT has not been investigated in NSCLC. A recent study was the first to analyse the relationships between the expression of SATB1 and EMT-associated proteins (E-cadherin, N-cadherin, SNAIL, SLUG, and Twist1) in NSCLC clinical samples. Moreover, it also investigated the impact of in vitro EMT induction in NSCLC cell cultures on SATB1 mRNA levels for the first time.

One of the main EMT hallmarks, usually observed at the very beginning of this process, is a decrease in the expression of E-cadherin, a calcium-dependent cell-adhesion molecule that is one of the key epithelial markers (69). E-cadherin mediates cell-to-cell junctions and interactions and prevents migration, therefore it is a known tumour suppressor protein (69,70). The loss of its expression not only results in the disruption of cell-to-cell contacts, but it was also demonstrated to activate downstream transcriptional pathways, leading to invasion and metastasis (71,72). In NSCLC, a decreased E-cadherin level was shown to be associated with poor tumour differentiation, the presence of lymph node metastasis, and an advanced stage of the disease (7376). In our study, we observed E-cadherin expression in most of the analysed NSCLC samples. However, there was no relationship with the patient clinicopathological or survival data. This supports previous findings by Grigoras et al and Myong et al, who assessed E-cadherin expression in NSCLC and were not able to observe any significant associations with the majority of the clinicopathological factors analysed or the patients' survival (77,78). However, although there are some studies in which the prognostic significance of the loss of E-cadherin expression in NSCLC has not been proven yet (73,7678), in the majority of the experiments, a decreased E-cadherin level was found to be a negative prognostic factor for NSCLC patients, something that has also been confirmed by meta-analyses (75,79,80).

Another protein from the cadherin family, closely associated with EMT progression, is N-cadherin, a key mesenchymal marker necessary for cell migration (69,70). While E-cadherin expression is being supressed during EMT, the N-cadherin level significantly increases in a process called ‘a cadherin switch’ (69,70,81). Aberrant N-cadherin expression has been observed in most of the epithelial-derived solid tumours, and it was shown to be a factor that influenced patient survival negatively and was related to a high tumour grade and the presence of lymph node metastasis (81,82). In the current study, although N-cadherin expression was only noted in ~8% of the NSCLC samples analysed, it was significantly related to the more advanced stage of the disease-both in the whole study cohort and in the LSCC subtype analysed separately. An increased N-cadherin level also seemed to be slightly associated with the LSCC histology and the presence of lymph node metastasis in LSCC tumours, but the results were on the verge of statistical significance (P=0.07, in both cases). Moreover, we observed that N-cadherin expression in LSCC tumours was significantly correlated with a shorter overall survival of the patients. Our results share a number of similarities with the findings of Hui et al, who also observed elevated N-cadherin scores in advanced stage tumours and the negative impact of N-cadherin expression on NSCLC patient prognosis (83). Analogous relationships were seen in nasopharyngeal and oral squamous cell carcinomas as well (84,85).

The whole EMT process is controlled at the transcriptional and post-transcriptional level by numerous transcription factors, chromatin modifying enzymes, miRNAs and long non-coding RNAs (911). The most important transcription regulators coordinating the shift from epithelial to mesenchymal phenotype are SNAIL, SLUG, and Twist1 (911). These proteins are responsible for downregulating E-cadherin and activating traits leading to the upregulation of mesenchymal markers, additionally influencing apoptosis, angiogenesis, and cell migration (8688). The overexpression of each of these transcription factors has been confirmed to have a negative prognostic significance in most common epithelial cancers (8890). In our study, we observed an elevated SNAIL, SLUG, and Twist1 immunoreactivity in NSCLC compared to NMLT samples, which is in line with previous literature reports (9193). However, some researchers noticed SNAIL staining in the nuclei of tumour cells (86,94), while we only observed it in the cytoplasm. These differences may be due to the use of different IHC methodologies and antibodies. However, recent reports suggest that subcellular SNAIL localization may be one of the mechanisms to regulate its activity during EMT (95), so this issue surely needs to be investigated more thoroughly in the future. In this study, we did not note any difference in SNAIL staining between the particular NSCLC subtypes, which is consistent with Merikallio et al (86) and Yanagawa et al (91). In LSCC, elevated SNAIL scores were associated with the presence of lymph node metastases and an advanced stage of the disease, which fits the results obtained by Abd El-Rehim et al (94). Although some studies stated that elevated SNAIL scores are related to a shortened NSCLC patient survival (86,91), in our research, SNAIL expression did not reach prognostic significance.

It is known that SLUG can promote lung cancer invasion and metastasis (96), but there are not many reports analysing its expression in NSCLC clinical samples. Therefore, it is challenging to discuss the results obtained in the current study. In 2014, Merikaillo et al investigated SLUG immunostaining in more than 250 NSCLC samples, but the only associations analysed were those with the histological subtype of the tumour and the patient survival (97). On the other hand, Hung et al analysed SLUG expression in 85 NSCLC tumours, but they did not observe any associations with the patient clinicopathological data (98). In our study, nuclear SLUG staining was significantly associated with the LSCC histology, whereas staining in the cytoplasm of the tumour cells did not differ among particular NSCLC subtypes, which is in good agreement with Merikallio et al (97). In LSCC, both nuclear and cytoplasmic SLUG expression was positively associated with the size of the tumour and the stage of the disease. In AC, an elevated cytoplasmic SLUG staining was related to a poor degree of tumour differentiation. In contrast to Jiang et al (93) and Merikallio et al (97), we did not observe any impact of SLUG expression on patient survival.

We observed Twist1 expression predominantly in LSCC tumours, which is consistent with Jiang et al (93). Additionally, its elevated scores were related to a more advanced stage of the disease, which supports the results obtained by Hui et al (83). However, a meta-analysis of 5 different studies regarding Twist1 expression in NSCLC did not confirm these findings, revealing that in the experiments analysed, Twist1 overexpression was associated only with the presence of lymph node metastasis (99). We did not find Twist1 expression to be prognostically significant in the NSCLC samples analysed. Although the meta-analyses (based mainly on Chinese population studies) stated that Twist1 overexpression was a negative prognostic factor (99,100) in NSCLC, there are also several studies that lend support to our findings (83,101).

The association between SATB1 level and the expression of various EMT-associated factors has been revealed, to date, in breast, colorectal, prostate, liver, and bladder cancers (43,45,46,52,55,56). Nevertheless, these studies were mostly based on the loss-of-function cell culture models and did not analyse the potential link between the expression of SATB1 and the EMT-promoting factors in clinical material. It is known that SATB1 immunostaining is correlated with the expression of vimentin and negatively correlated with the expression of E-cadherin and CK20 in colorectal cancer clinical samples (51), but no reports concerning lung carcinomas are available. Therefore, we are the first researchers to investigate the relationship between the expression of SATB1 and EMT-associated proteins in NSCLC tumours. We found SNAIL, SLUG, and Twist1 expression levels to be positively correlated with SATB1 scores in the whole study cohort, which clearly indicates a possible association between SATB1 expression and the progression of EMT in these tumours. Moreover, nuclear and cytoplasmic SLUG staining was positively correlated with the expression of SATB1 in particular NSCLC subtypes as well. Surprisingly, the associations were the strongest in LSCC samples, which seems to be contradictory with the positive prognostic significance of SATB1 expression in these tumours observed by Selinger et al (58). However, in our current and recent studies, we have not noted a statistically significant link between either SATB1 or SLUG expression and LSCC patient survival (59). In the currently analysed material, we also observed a positive correlation between SATB1 and Twist1 expressions in AC samples, which is consistent with the postulated tumour-promoting role of these two factors in adenocarcinomas (60,102).

Although SATB1 has been shown to have an impact on E-cadherin and N-cadherin expressions in numerous cancer cell lines (43,45,46,52,55,56), we did not note any relationship among the levels of these factors in NSCLC clinical samples. Instead, surprisingly, we observed a significant positive correlation between E-cadherin expression and the Ki-67 proliferative index, both in the whole study cohort and in the AC subtype alone. Similar associations have been reported in laryngeal and endometrial cancers (103,104), but not in NSCLC. N-cadherin, SNAIL, SLUG, and Twist1 scores were also positively correlated with the Ki-67 proliferative index, which seems to be consistent with their tumour-promoting function.

It has been revealed in several studies that ectopic SATB1 overexpression is enough to induce EMT-like changes in cancer cells. However, there are no described experiments in which the reverse relationship would be studied (the impact of EMT on SATB1 level). We were the first ones to investigate whether the induction of EMT in cultured NSCLC cells affects the expression of SATB1 somehow. We revealed that the SATB1 mRNA level significantly increased after TGF-β1 exposure, both in the A549 (AC) and the NCI-H1703 (LSCC) cell lines. These changes in SATB1 expression were accompanied by a visible morphological shift from an epithelial to a mesenchymal phenotype, together with a significant increase in the expression of genes coding for N-cadherin and EMT-promoting transcription factors (CDH2, SNAI1, and SNAI2, respectively). These results may indicate the role of SATB1 as an EMT-inducer in NSCLC.

In conclusion, SATB1 is a known tumour-promoting factor whose negative prognostic significance has been observed in numerous cancers. It has been shown to be associated with an invasive tumour phenotype and to promote metastatic spread. In recent years, an increasing number of reports have also pointed to a possible role of SATB1 as an EMT-promoting factor. Links between SATB1 expression and the progression of EMT have been noticed in several epithelial cancers, including those of the breast, colon, liver, prostate, and bladder. However, non-small cell lung carcinomas, the most abundant lung cancer subtype, have never be analysed in the context of SATB1/EMT associations. We were the first to analyse the relationship between the expression of SATB1 and key EMT-associated proteins (E-cadherin, N-cadherin, SNAIL, SLUG, and Twist1) in NSCLC clinical samples. Additionally, we also investigated the impact of EMT induction in NSCLC cell lines on the expression of SATB1 mRNA. We observed significant positive correlations between the expression of SATB1 and the most crucial EMT-promoting transcription factors: SNAIL, SLUG, and Twist1. We also showed that SATB1 expression significantly increased after in vitro EMT induction in the A549 and the NCI-H1703 NSCLC cell lines. These results may indicate the role of SATB1 as one of the positive EMT regulators in NSCLC.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

This study was funded by a grant from the Medical University in Wroclaw, Poland (no. STM.A110.17.023).

Availability of data and materials

The datasets analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

Conceptualization of the research study was achieved by NGP and PD. Formal analysis was conducted by NGP. Funding acquisition was by NGP. Investigation and experimentation was performed by NGP and AP. Methodology was designed by NGP and AP. Resources were the responsibility of AR and supervision was conducted by MPO and PD. Visualization was performed by NGP. Writing of the original draft was performed by NGP and writing, review and editing was conducted by AR, MPO and PD. All authors read and approved the manuscript and agree to be accountable for all aspects of the research in ensuring that the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Ethics approval and consent to participate

The present study was approved by the Bioethics Commission at the Wroclaw Medical University in Poland (approval no. KB-632/2017). The present study was conducted on archival clinical material only, thus no patient consent was needed.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Ferlay J, Ervik M, Lam F, Colombet M, Mery L, Piñeros M, Znaor A, Soerjomataram I and Bray F: Global Cancer Observatory: Cancer Today. Int Agency Res Cancer. 2018 https://publications.iarc.fr/Databases/Iarc-Cancerbases/Cancer-Today-Powered-By-GLOBOCAN-2018--2018December 62019 12 06;

2 

Zappa C and Mousa SA: Non-small cell lung cancer: Current treatment and future advances. Transl Lung Cancer Res. 5:288–300. 2016. View Article : Google Scholar : PubMed/NCBI

3 

Travis WD, Brambilla E, Burke AP, Marx A and Nicholson AG: Introduction to The 2015 World Health Organization Classification of tumours of the lung, pleura, thymus and heart. J Thorac Oncol. 10:1240–1242. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Perlikos F, Harrington KJ and Syrigos KN: Key molecular mechanisms in lung cancer invasion and metastasis: A comprehensive review. Crit Rev Oncol Hematol. 87:1–11. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Tamura T, Kurishima K, Nakazawa K, Kagahashi K, Ishikawa H, Satoh H and Hizawa N: Specific organ metastases and survival in metastatic non-small-cell lung cancer. Mol Clin Oncol. 3:217–221. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Milovanovic IS, Stjepanovic M and Mitrovic D: Distribution patterns of the metastases of the lung carcinoma in relation to histological type of the primary tumor: An autopsy study. Ann Thorac Med. 12:191–198. 2017. View Article : Google Scholar : PubMed/NCBI

7 

Hsu F, De Caluwe A, Anderson D, Nichol A, Toriumi T and Ho C: Patterns of spread and prognostic implications of lung cancer metastasis in an era of driver mutations. Curr Oncol. 24:228–233. 2017. View Article : Google Scholar : PubMed/NCBI

8 

Howlader N, Noone AM, Krapcho M, Miller D, Brest A, Yu M, Ruhl J, Tatalovich Z, Mariotto A, Lewis DR, et al: SEER Cancer Statistics Review, 1975–2017. https://seer.cancer.gov/csr/1975_2017April 15–2020

9 

Diepenbruck M and Christofori G: Epithelial-mesenchymal transition (EMT) and metastasis: Yes, no, maybe? Curr Opin Cell Biol. 43:7–13. 2016. View Article : Google Scholar : PubMed/NCBI

10 

Yang X, Liang X, Zheng M and Tang Y: Cellular phenotype plasticity in cancer dormancy and metastasis. Front Oncol. 8:5052018. View Article : Google Scholar : PubMed/NCBI

11 

De Craene B and Berx G: Regulatory networks defining EMT during cancer initiation and progression. Nat Rev Cancer. 13:97–110. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Zhang Y and Weinberg RA: Epithelial-to-mesenchymal transition in cancer: Complexity and opportunities. Front Med. 12:361–373. 2018. View Article : Google Scholar : PubMed/NCBI

13 

Santamaria PG, Moreno-Bueno G, Portillo F and Cano A: EMT: Present and future in clinical oncology. Mol Oncol. 11:718–738. 2017. View Article : Google Scholar : PubMed/NCBI

14 

Singh M, Yelle N, Venugopal C and Singh SK: EMT: Mechanisms and therapeutic implications. Pharmacol Ther. 182:80–94. 2018. View Article : Google Scholar : PubMed/NCBI

15 

Pastushenko I and Blanpain C: EMT transition states during tumor progression and metastasis. Trends Cell Biol. 29:212–226. 2019. View Article : Google Scholar : PubMed/NCBI

16 

Frixen UH, Behrens J, Sachs M, Eberle G, Voss B, Warda A, Löchner D and Birchmeier W: E-cadherin-mediated cell-cell adhesion prevents invasiveness of human carcinoma cells. J Cell Biol. 113:173–185. 1991. View Article : Google Scholar : PubMed/NCBI

17 

Thiery JP: Epithelial-mesenchymal transitions in tumour progression. Nat Rev Cancer. 2:442–454. 2002. View Article : Google Scholar : PubMed/NCBI

18 

Nagathihalli NS, Massion PP, Gonzalez AL, Lu P and Datta PK: Smoking induces epithelial-to-mesenchymal transition in non-small cell lung cancer through HDAC-mediated downregulation of E-cadherin. Mol Cancer Ther. 11:2362–2372. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Mahmood MQ, Walters EH, Shukla SD, Weston S, Muller HK, Ward C and Sohal SS: β-catenin, Twist and Snail: Transcriptional regulation of EMT in smokers and COPD, and relation to airflow obstruction. Sci Rep. 7:108322017. View Article : Google Scholar : PubMed/NCBI

20 

Zhu X, Chen L, Liu L and Niu X: EMT-Mediated Acquired EGFR-TKI resistance in NSCLC: Mechanisms and strategies. Front Oncol. 9:10442019. View Article : Google Scholar : PubMed/NCBI

21 

Tulchinsky E, Demidov O, Kriajevska M, Barlev NA and Imyanitov E: EMT: A mechanism for escape from EGFR-targeted therapy in lung cancer. Biochim Biophys Acta Rev Cancer. 1871:29–39. 2019. View Article : Google Scholar : PubMed/NCBI

22 

Bronte G, Bravaccini S, Bronte E, Burgio MA, Rolfo C, Delmonte A and Crinò L: Epithelial-to-mesenchymal transition in the context of epidermal growth factor receptor inhibition in non-small-cell lung cancer. Biol Rev Camb Philos Soc. 93:1735–1746. 2018. View Article : Google Scholar : PubMed/NCBI

23 

Li M, Yang J, Zhou W, Ren Y, Wang X, Chen H, Zhang J, Chen J, Sun Y, Cui L, et al: Activation of an AKT/FOXM1/STMN1 pathway drives resistance to tyrosine kinase inhibitors in lung cancer. Br J Cancer. 117:974–983. 2017. View Article : Google Scholar : PubMed/NCBI

24 

Santamaría PG, Moreno-Bueno G and Cano A: Contribution of epithelial plasticity to therapy resistance. J Clin Med. 8:6762019. View Article : Google Scholar

25 

Liang SQ, Marti TM, Dorn P, Froment L, Hall SR, Berezowska S, Kocher G, Schmid RA and Peng RW: Blocking the epithelial-to-mesenchymal transition pathway abrogates resistance to anti-folate chemotherapy in lung cancer. Cell Death Dis. 6:e18242015. View Article : Google Scholar : PubMed/NCBI

26 

Zheng HC: The molecular mechanisms of chemoresistance in cancers. Oncotarget. 8:59950–59964. 2017. View Article : Google Scholar : PubMed/NCBI

27 

Fischer KR, Durrans A, Lee S, Sheng J, Li F, Wong ST, Choi H, El Rayes T, Ryu S, Troeger J, et al: Epithelial-to-mesenchymal transition is not required for lung metastasis but contributes to chemoresistance. Nature. 527:472–476. 2015. View Article : Google Scholar : PubMed/NCBI

28 

Kohwi-Shigematsu T, Kohwi Y, Takahashi K, Richards HW, Ayers SD, Han HJ and Cai S: SATB1-mediated functional packaging of chromatin into loops. Methods. 58:243–254. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Galande S, Purbey PK, Notani D and Kumar PP: The third dimension of gene regulation: Organization of dynamic chromatin loopscape by SATB1. Curr Opin Genet Dev. 17:408–414. 2007. View Article : Google Scholar : PubMed/NCBI

30 

Kohwi-Shigematsu T, Poterlowicz K, Ordinario E, Han HJ, Botchkarev VA and Kohwi Y: Genome organizing function of SATB1 in tumor progression. Semin Cancer Biol. 23:72–79. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Fessing MY, Mardaryev AN, Gdula MR, Sharov AA, Sharova TY, Rapisarda V, Gordon KB, Smorodchenko AD, Poterlowicz K, Ferone G, et al: p63 regulates Satb1 to control tissue-specific chromatin remodeling during development of the epidermis. J Cell Biol. 194:825–839. 2011. View Article : Google Scholar : PubMed/NCBI

32 

Pavan Kumar P, Purbey PK, Sinha CK, Notani D, Limaye A, Jayani RS and Galande S: Phosphorylation of SATB1, a global gene regulator, acts as a molecular switch regulating its transcriptional activity in vivo. Mol Cell. 22:231–243. 2006. View Article : Google Scholar : PubMed/NCBI

33 

Purbey PK, Singh S, Notani D, Kumar PP, Limaye AS and Galande S: Acetylation-dependent interaction of SATB1 and CtBP1 mediates transcriptional repression by SATB1. Mol Cell Biol. 29:1321–1337. 2009. View Article : Google Scholar : PubMed/NCBI

34 

Nagpal N, Ahmad HM, Molparia B and Kulshreshtha R: MicroRNA-191, an estrogen-responsive microRNA, functions as an oncogenic regulator in human breast cancer. Carcinogenesis. 34:1889–1899. 2013. View Article : Google Scholar : PubMed/NCBI

35 

Brown CY, Dayan S, Wong SW, Kaczmarek A, Hope CM, Pederson SM, Arnet V, Goodall GJ, Russell D, Sadlon TJ and Barry SC: FOXP3 and miR-155 cooperate to control the invasive potential of human breast cancer cells by down regulating ZEB2 independently of ZEB1. Oncotarget. 9:27708–27727. 2018. View Article : Google Scholar : PubMed/NCBI

36 

Li QQ, Chen ZQ, Cao XX, Xu JD, Xu JW, Chen YY, Wang WJ, Chen Q, Tang F, Liu XP and Xu ZD: Involvement of NF-κB/miR-448 regulatory feedback loop in chemotherapy-induced epithelial-mesenchymal transition of breast cancer cells. Cell Death Differ. 18:16–25. 2011. View Article : Google Scholar : PubMed/NCBI

37 

McInnes N, Sadlon TJ, Brown CY, Pederson S, Beyer M, Schultze JL, McColl S, Goodall GJ and Barry SC: FOXP3 and FOXP3-regulated microRNAs suppress SATB1 in breast cancer cells. Oncogene. 31:1045–1054. 2012. View Article : Google Scholar : PubMed/NCBI

38 

Pan X, Li D, Huo J, Kong F, Yang H and Ma X: LINC01016 promotes the malignant phenotype of endometrial cancer cells by regulating the miR-302a-3p/miR-3130-3p/NFYA/SATB1 axis. Cell Death Dis. 9:3032018. View Article : Google Scholar : PubMed/NCBI

39 

Lopes-Ramos CM, Habr-Gama A, Quevedo Bde S, Felício NM, Bettoni F, Koyama FC, Asprino PF, Galante PA, Gama-Rodrigues J, Camargo AA, et al: Overexpression of miR-21-5p as a predictive marker for complete tumor regression to neoadjuvant chemoradiotherapy in rectal cancer patients. BMC Med Genomics. 7:682014. View Article : Google Scholar : PubMed/NCBI

40 

Kowalczyk AE, Krazinski BE, Godlewski J, Grzegrzolka J, Kiewisz J, Kwiatkowski P, Sliwinska-Jewsiewicka A, Dziegiel P and Kmiec Z: SATB1 is Down-regulated in clear cell renal cell carcinoma and correlates with miR-21-5p overexpression and poor prognosis. Cancer Genomics Proteomics. 13:209–217. 2016.PubMed/NCBI

41 

Alvarez JD, Yasui DH, Niida H, Joh T, Loh DY and Kohwi-Shigematsu T: The MAR-binding protein SATB1 orchestrates temporal and spatial expression of multiple genes during T-cell development. Genes Dev. 14:521–535. 2000.PubMed/NCBI

42 

Savarese F, Dávila A, Nechanitzky R, De La Rosa-Velazquez I, Pereira CF, Engelke R, Takahashi K, Jenuwein T, Kohwi-Shigematsu T, Fisher AG and Grosschedl R: Satb1 and Satb2 regulate embryonic stem cell differentiation and Nanog expression. Genes Dev. 23:2625–2638. 2009. View Article : Google Scholar : PubMed/NCBI

43 

Han HJ, Russo J, Kohwi Y and Kohwi-Shigematsu T: SATB1 reprogrammes gene expression to promote breast tumour growth and metastasis. Nature. 452:187–193. 2008. View Article : Google Scholar : PubMed/NCBI

44 

Nodin B, Johannesson H, Wangefjord S, O'Connor DP, Lindquist KE, Uhlén M, Jirström K and Eberhard J: Molecular correlates and prognostic significance of SATB1 expression in colorectal cancer. Diagn Pathol. 7:1152012. View Article : Google Scholar : PubMed/NCBI

45 

Shukla S, Sharma H, Abbas A, MacLennan GT, Fu P, Danielpour D and Gupta S: Upregulation of SATB1 is associated with prostate cancer aggressiveness and disease progression. PLoS One. 8:e535272013. View Article : Google Scholar : PubMed/NCBI

46 

Tu W, Luo M, Wang Z, Yan W, Xia Y, Deng H, He J, Han P and Tian D: Upregulation of SATB1 promotes tumor growth and metastasis in liver cancer. Liver Int. 32:1064–1078. 2012. View Article : Google Scholar : PubMed/NCBI

47 

Han B, Luan L, Xu Z and Wu B: Expression and biological roles of SATB1 in human bladder cancer. Tumor Biol. 34:2943–2949. 2013. View Article : Google Scholar : PubMed/NCBI

48 

Nodin B, Hedner C, Uhlén M and Jirström K: Expression of the global regulator SATB1 is an independent factor of poor prognosis in high grade epithelial ovarian cancer. J Ovarian Res. 5:242012. View Article : Google Scholar : PubMed/NCBI

49 

Wang S, Zeng J, Xiao R, Xu G, Liu G, Xiong D, Ye Y, Chen B, Wang H, Luo Q and Huang Z: Poor prognosis and SATB1 overexpression in solid tumors: A meta-analysis. Cancer Manag Res. 10:1471–1478. 2018. View Article : Google Scholar : PubMed/NCBI

50 

Sun Z, Zhang C, Zou X, Jiang G, Xu Z, Li W and Xie H: Special AT-rich sequence-binding protein-1 participates in the maintenance of breast cancer stem cells through regulation of the Notch signaling pathway and expression of Snail1 and Twist1. Mol Med Rep. 11:3235–3242. 2015. View Article : Google Scholar : PubMed/NCBI

51 

Lv JH, Wang F, Shen MH, Wang X and Zhou XJ: SATB1 expression is correlated with β-catenin associated epithelial-mesenchymal transition in colorectal cancer. Cancer Biol Ther. 17:254–261. 2016. View Article : Google Scholar : PubMed/NCBI

52 

Frömberg A, Rabe M and Aigner A: Multiple effects of the special AT-rich binding protein 1 (SATB1) in colon carcinoma. Int J Cancer. 135:2537–2546. 2014. View Article : Google Scholar

53 

Mao L, Yang C, Wang J, Li W, Wen R, Chen J and Zheng J: SATB1 is overexpressed in metastatic prostate cancer and promotes prostate cancer cell growth and invasion. J Transl Med. 11:1112013. View Article : Google Scholar : PubMed/NCBI

54 

Mao LJ, Zhang J, Liu N, Fan L, Yang DR, Xue BX, Shan YX and Zheng JN: Oncolytic virus carrying shRNA targeting SATB1 inhibits prostate cancer growth and metastasis. Tumor Biol. 36:9073–9081. 2015. View Article : Google Scholar : PubMed/NCBI

55 

Qi H, Fu X, Li Y, Pang X, Chen S, Zhu X, Li F and Tan W: SATB1 promotes epithelial-mesenchymal transition and metastasis in prostate cancer. Oncol Lett. 13:2577–2582. 2017. View Article : Google Scholar : PubMed/NCBI

56 

Wan F, Cheng C, Wang Z, Xiao X, Zeng H, Xing S, Chen X, Wang J, Li S, Zhang Y, et al: SATB1 overexpression regulates the development and progression in bladder cancer through EMT. PLoS One. 10:e01175182015. View Article : Google Scholar : PubMed/NCBI

57 

Baguma-Nibasheka M, Angka HE, Inanlou MR and Kablar B: Microarray analysis of Myf5-/-:MyoD-/-hypoplastic mouse lungs reveals a profile of genes involved in pneumocyte differentiation. Histol Histopathol. 22:483–495. 2007.PubMed/NCBI

58 

Selinger CI, Cooper WA, Al-Sohaily S, Mladenova DN, Pangon L, Kennedy CW, McCaughan BC, Stirzaker C and Kohonen-Corish MR: Loss of special AT-rich binding protein 1 expression is a marker of poor survival in lung cancer. J Thorac Oncol. 6:1179–1189. 2011. View Article : Google Scholar : PubMed/NCBI

59 

Glatzel-Plucinska N, Piotrowska A, Grzegrzolka J, Olbromski M, Rzechonek A, Dziegiel P and Podhorska-Okolow M: SATB1 level correlates with Ki-67 expression and is a positive prognostic factor in non-small cell lung carcinoma. Anticancer Res. 38:723–736. 2018.PubMed/NCBI

60 

Huang B, Zhou H, Wang S, Lang XP and Wang X: Effect of silencing SATB1 on proliferation, invasion and apoptosis of A549 human lung adenocarcinoma cells. Oncol Lett. 12:3818–3824. 2016. View Article : Google Scholar : PubMed/NCBI

61 

Huang B, Zhou H, Wang X and Liu Z: Silencing SATB1 with siRNA inhibits the proliferation and invasion of small cell lung cancer cells. Cancer Cell Int. 13:82013. View Article : Google Scholar : PubMed/NCBI

62 

Detterbeck FC, Boffa DJ and Tanoue LT: The new lung cancer staging system. Chest. 136:260–271. 2009. View Article : Google Scholar : PubMed/NCBI

63 

Tuffaha MSA: Phenotypic and Genotypic Diagnosis of Malignancies: An Immunohistochemical and Molecular Approach. 1st edition. Wiley-VCH; 2008, View Article : Google Scholar

64 

Mogilner A and Keren K: The shape of motile cells. Curr Biol. 19:R762–R771. 2009. View Article : Google Scholar : PubMed/NCBI

65 

Glatzel-Plucinska N, Piotrowska A, Dziegiel P and Podhorska-Okołów M: The role of SATB1 in tumour progression and metastasis. Int J Mol Sci. 20:41562019. View Article : Google Scholar : PubMed/NCBI

66 

Zhang J, Zhang B, Zhang X, Sun Y, Wei X, McNutt MA, Lu S, Liu Y, Zhang D, Wang M, et al: SATB1 expression is associated with biologic behavior in colorectal carcinoma in vitro and in vivo. PLoS One. 8:e479022013. View Article : Google Scholar : PubMed/NCBI

67 

Mansour MA, Hyodo T, Akter KA, Kokuryo T, Uehara K, Nagino M and Senga T: SATB1 and SATB2 play opposing roles in c-Myc expression and progression of colorectal cancer. Oncotarget. 7:4993–5006. 2016. View Article : Google Scholar : PubMed/NCBI

68 

Zhang Y, Tian X, Ji H, Guan X, Xu W, Dong B, Zhao M, Wei M, Ye C, Sun Y, et al: Expression of SATB1 promotes the growth and metastasis of colorectal cancer. PLoS One. 9:e1004132014. View Article : Google Scholar : PubMed/NCBI

69 

Loh CY, Chai JY, Tang TF, Wong WF, Sethi G, Shanmugam MK, Chong PP and Looi CY: The E-Cadherin and N-Cadherin switch in epithelial-to-mesenchymal transition: Signaling, therapeutic implications, and challenges. Cells. 8:11182019. View Article : Google Scholar : PubMed/NCBI

70 

Berx G and van Roy F: Involvement of members of the cadherin superfamily in cancer. Cold Spring Harb Perspect Biol. 1:a0031292009. View Article : Google Scholar : PubMed/NCBI

71 

Bae GY, Choi SJ, Lee JS, Jo J, Lee J, Kim J and Cha HJ: Loss of E-cadherin activates EGFR-MEK/ERK signaling, which promotes invasion via the ZEB1/MMP2 axis in non-small cell lung cancer. Oncotarget. 4:2512–2522. 2013. View Article : Google Scholar : PubMed/NCBI

72 

Onder TT, Gupta PB, Mani SA, Yang J, Lander ES and Weinberg RA: Loss of E-Cadherin promotes metastasis via multiple downstream transcriptional pathways. Cancer Res. 68:3645–3654. 2008. View Article : Google Scholar : PubMed/NCBI

73 

Kase S, Sugio K, Yamazaki K, Okamoto T, Yano T and Sugimachi K: Expression of E-cadherin and beta-catenin in human non-small cell lung cancer and the clinical significance. Clin Cancer Res. 6:4789–4796. 2000.PubMed/NCBI

74 

Bremnes RM, Veve R, Gabrielson E, Hirsch FR, Baron A, Bemis L, Gemmill RM, Drabkin HA and Franklin WA: High-throughput tissue microarray analysis used to evaluate biology and prognostic significance of the E-cadherin pathway in non-small-cell lung cancer. J Clin Oncol. 20:2417–2428. 2002. View Article : Google Scholar : PubMed/NCBI

75 

Yan B, Zhang W, Jiang LY, Qin WX and Wang X: Reduced E-Cadherin expression is a prognostic biomarker of non-small cell lung cancer: A meta-analysis based on 2395 subjects. Int J Clin Exp Med. 7:4352–4356. 2014.PubMed/NCBI

76 

Lee YC, Wu CT, Chen CS and Chang YL: E-Cadherin expression in surgically-resected non-small cell lung cancers-A clinicopathological study. Thorac Cardiovasc Surg. 48:294–299. 2000. View Article : Google Scholar : PubMed/NCBI

77 

Grigoras ML, Arghirescu TS, Folescu R, Talpoş IC, Gîndac CM, Zamfir CL, Cornianu M, Anghel MD and Levai CM: Expression of E-cadherin in lung carcinoma, other than those with small cells (NSCLC). Rom J Morphol Embryol. 58:1317–1325. 2017.PubMed/NCBI

78 

Myong NH: Reduced expression of E-cadherin in human non-small cell lung carcinoma. Cancer Res Treat. 36:56–61. 2004. View Article : Google Scholar : PubMed/NCBI

79 

Wu Y, Liu HB, Ding M, Liu JN, Zhan P, Fu XS and Lu G: The impact of E-cadherin expression on non-small cell lung cancer survival: A meta-analysis. Mol Biol Rep. 39:9621–9628. 2012. View Article : Google Scholar : PubMed/NCBI

80 

Yang YL, Chen MW and Xian L: Prognostic and clinicopathological significance of downregulated E-cadherin expression in patients with non-small cell lung cancer (NSCLC): A meta-analysis. PLoS One. 9:e997632014. View Article : Google Scholar : PubMed/NCBI

81 

Mrozik KM, Blaschuk OW, Cheong CM, Zannettino ACW and Vandyke K: N-cadherin in cancer metastasis, its emerging role in haematological malignancies and potential as a therapeutic target in cancer. BMC Cancer. 18:9392018. View Article : Google Scholar : PubMed/NCBI

82 

Luo Y, Yu T, Zhang Q, Fu Q, Hu Y, Xiang M, Peng H, Zheng T, Lu L and Shi H: Upregulated N-cadherin expression is associated with poor prognosis in epithelial-derived solid tumours: A meta-analysis. Eur J Clin Invest. 48:e129032018. View Article : Google Scholar : PubMed/NCBI

83 

Hui L, Zhang S, Dong X, Tian D, Cui Z and Qiu X: Prognostic significance of twist and N-cadherin expression in NSCLC. PLoS One. 8:e621712013. View Article : Google Scholar : PubMed/NCBI

84 

Sun H, Liu M, Wu X, Yang C, Zhang Y, Xu Z, Gao K and Wang F: Overexpression of N-cadherin and β-catenin correlates with poor prognosis in patients with nasopharyngeal carcinoma. Oncol Lett. 13:1725–1730. 2017. View Article : Google Scholar : PubMed/NCBI

85 

Ozaki-Honda Y, Seki S, Fujiwara M, Matsuura M, Fujita S, Ikeda H, Umeda M, Ayuse T and Ikeda T: Prognostic prediction of oral squamous cell carcinoma by E-Cadherin and N-Cadherin expression in overall cells in tumor nests or tumor cells at the invasive front. Cancer Microenviron. 10:87–94. 2017. View Article : Google Scholar : PubMed/NCBI

86 

Merikallio H, Turpeenniemi-Hujanen T, Pääkkö P, Mäkitaro R, Riitta K, Salo S, Salo T, Harju T and Soini Y: Snail promotes an invasive phenotype in lung carcinoma. Respir Res. 13:1042012. View Article : Google Scholar : PubMed/NCBI

87 

Alves CC, Carneiro F, Hoefler H and Becker KF: Role of the epithelial-mesenchymal transition regulator Slug in primary human cancers. Front Biosci (Landmark Ed). 14:3035–3050. 2009. View Article : Google Scholar : PubMed/NCBI

88 

Zhao Z, Rahman MA, Chen ZG and Shin DM: Multiple biological functions of Twist1 in various cancers. Oncotarget. 8:20380–20393. 2017. View Article : Google Scholar : PubMed/NCBI

89 

Zhang P, Hu P, Shen H, Yu J, Liu Q and Du J: Prognostic role of Twist or Snail in various carcinomas: A systematic review and meta-analysis. Eur J Clin Invest. 44:1072–1094. 2014. View Article : Google Scholar : PubMed/NCBI

90 

Huang C, Zhang P, Zhang D and Weng X: The prognostic implication of slug in all tumour patients-a systematic meta-analysis. Eur J Clin Invest. 46:398–407. 2016. View Article : Google Scholar : PubMed/NCBI

91 

Yanagawa J, Walser TC, Zhu LX, Hong L, Fishbein MC, Mah V, Chia D, Goodglick L, Elashoff DA, Luo J, et al: Snail promotes CXCR2 ligand-dependent tumor progression in non-small cell lung carcinoma. Clin Cancer Res. 15:6820–6829. 2009. View Article : Google Scholar : PubMed/NCBI

92 

Liu CW, Li CH, Peng YJ, Cheng YW, Chen HW, Liao PL, Kang JJ and Yeng MH: Snail regulates Nanog status during the epithelial-mesenchymal transition via the Smad1/Akt/GSK3β signaling pathway in non-small-cell lung cancer. Oncotarget. 5:3880–3894. 2014. View Article : Google Scholar : PubMed/NCBI

93 

Jiang W, Pang XG, Wang Q, Shen YX, Chen XK and Xi JJ: Prognostic role of Twist, Slug, and Foxc2 expression in stage i non-small-cell lung cancer after curative resection. Clin Lung Cancer. 13:280–287. 2012. View Article : Google Scholar : PubMed/NCBI

94 

Abd El-Rehim DM, Abd-Elghany MI and Nazmy MH: Integrin-Linked kinase, Snail and multidrug resistance protein 1: Three concordant players in the progression of non-small cell lung cancer. J Egypt Natl Canc Inst. 27:129–137. 2015. View Article : Google Scholar : PubMed/NCBI

95 

Muqbil I, Wu J, Aboukameel A, Mohammad RM and Azmi AS: Snail nuclear transport: The gateways regulating epithelial-to-mesenchymal transition? Semin Cancer Biol. 27:39–45. 2014. View Article : Google Scholar : PubMed/NCBI

96 

Shih JY and Yang PC: The EMT regulator slug and lung carcinogenesis. Carcinogenesis. 32:1299–1304. 2011. View Article : Google Scholar : PubMed/NCBI

97 

Merikallio H, Turpeenniemi-Hujanen TT, Pääkkö P, Mäkitaro R, Kaarteenaho R, Lehtonen S, Salo S, Salo T, Harju T and Soini Y: Slug is associated with poor survival in squamous cell carcinoma of the lung. Int J Clin Exp Pathol. 7:5846–5854. 2014.PubMed/NCBI

98 

Hung PF, Hong TM, Chang CC, Hung CL, Hsu YL, Chang YL, Wu CT, Chang GC, Chan NL, Yu SL, et al: Hypoxia-induced Slug SUMOylation enhances lung cancer metastasis. J Exp Clin Cancer Res. 38:52019. View Article : Google Scholar : PubMed/NCBI

99 

Li M, Zhang X, Xu X, Wu J, Hu K, Guo X and Zhang P: Clinicopathological and prognostic significance of Twist overexpression in NSCLC. Oncotarget. 9:14642–14651. 2018. View Article : Google Scholar : PubMed/NCBI

100 

Zeng J, Zhan P, Wu G, Yang W, Liang W, Lv T and Song Y: Prognostic value of twist in lung cancer: Systematic review and meta-analysis. Transl Lung Cancer Res. 4:236–241. 2015.PubMed/NCBI

101 

Shi Y, Wu H, Zhang M, Ding L, Meng F and Fan X: Expression of the epithelial-mesenchymal transition-related proteins and their clinical significance in lung adenocarcinoma. Diagn Pathol. 8:892013. View Article : Google Scholar : PubMed/NCBI

102 

Pallier K, Cessot A, Côté JF, Just PA, Cazes A, Fabre E, Danel C, Riquet M, Devouassoux-Shisheboran M, Ansieau S, et al: TWIST1 a new determinant of epithelial to mesenchymal transition in EGFR mutated lung adenocarcinoma. PLoS One. 7:e299542012. View Article : Google Scholar : PubMed/NCBI

103 

Barutçu O, Kara M, Muratlı A, Güçlü O and Dereköy S: Clinical significance of Ki-67, c-erbB-2 and E-cadherin expressions in open partial laryngectomy patients. Kulak Burun Bogaz Ihtis Derg. 26:283–292. 2016. View Article : Google Scholar

104 

González-Rodilla I, Aller L, Llorca J, Muñoz AB, Verna V, Estévez J and Schneider J: The E-Cadherin expression vs. tumor cell proliferation paradox in endometrial cancer. Anticancer Res. 33:5091–5095. 2013.

Related Articles

Journal Cover

June-2021
Volume 45 Issue 6

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Glatzel‑Plucinska N, Piotrowska A, Rzechonek A, Podhorska‑Okolow M and Dziegiel P: SATB1 protein is associated with the epithelial‑mesenchymal transition process in non‑small cell lung cancers. Oncol Rep 45: 118, 2021
APA
Glatzel‑Plucinska, N., Piotrowska, A., Rzechonek, A., Podhorska‑Okolow, M., & Dziegiel, P. (2021). SATB1 protein is associated with the epithelial‑mesenchymal transition process in non‑small cell lung cancers. Oncology Reports, 45, 118. https://doi.org/10.3892/or.2021.8069
MLA
Glatzel‑Plucinska, N., Piotrowska, A., Rzechonek, A., Podhorska‑Okolow, M., Dziegiel, P."SATB1 protein is associated with the epithelial‑mesenchymal transition process in non‑small cell lung cancers". Oncology Reports 45.6 (2021): 118.
Chicago
Glatzel‑Plucinska, N., Piotrowska, A., Rzechonek, A., Podhorska‑Okolow, M., Dziegiel, P."SATB1 protein is associated with the epithelial‑mesenchymal transition process in non‑small cell lung cancers". Oncology Reports 45, no. 6 (2021): 118. https://doi.org/10.3892/or.2021.8069